ArticlePDF Available

Glucose Stimulates Translocation of the Homeodomain Transcription Factor PDX1 from the Cytoplasm to the Nucleus in Pancreatic -Cells

Authors:

Abstract and Figures

One of the mechanisms whereby glucose stimulates insulin gene transcription in pancreatic β-cells involves activation of the homeodomain transcription factor PDX1 (pancreatic/duodenal homeobox-1) via a stress-activated pathway involving stress-activated protein kinase 2 (SAPK2, also termed RK/p38, CSBP, and Mxi2). In the present study we show, by Western blotting and electrophoretic mobility shift assay, that in human islets of Langerhans incubated in low glucose (3 mm) PDX1 exists as an inactive 31-kDa protein localized exclusively in the cytoplasm. Transfer of the islets to high (16 mm) glucose results in rapid (within 10 min) conversion of PDX1 to an active 46-kDa form that was present predominantly in the nucleus. Activation of PDX1 appears to involve phosphorylation, as shown by incorporation of32Pi into the 46-kDa form of the protein. These effects of glucose could be mimicked by chemical stress (sodium arsenite), or by overexpression of SAPK2 in the β-cell line MIN6. Overexpression of SAPK2 also stimulated PDX1-dependent transcription of a –50 to –250 region of the human insulin gene promoter linked to a firefly luciferase reporter gene. The effects of glucose were inhibited by the SAPK2 inhibitor SB 203580, and by wortmannin and LY 294002, which inhibit phosphatidylinositol 3-kinase, although the effects of stress (arsenite) were inhibited only by SB 203580. These results demonstrate that glucose regulates the insulin gene promoter through activation and nuclear translocation of PDX1 via the SAPK2 pathway.
Content may be subject to copyright.
Glucose Stimulates Translocation of the Homeodomain
Transcription Factor PDX1 from the Cytoplasm to the
Nucleus in Pancreatic
b
-Cells*
(Received for publication, June 11, 1998, and in revised form, October 7, 1998)
Wendy M. Macfarlane‡, Caroline M. McKinnon‡, Zoe A. Felton-Edkins‡, Helen Cragg‡,
Roger F. L. James§, and Kevin Docherty‡
From the Department of Molecular and Cell Biology, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD and
the §Department of Surgery, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, United Kingdom
One of the mechanisms whereby glucose stimulates
insulin gene transcription in pancreatic
b
-cells involves
activation of the homeodomain transcription factor
PDX1 (pancreatic/duodenal homeobox-1) via a stress-
activated pathway involving stress-activated protein ki-
nase 2 (SAPK2, also termed RK/p38, CSBP, and Mxi2). In
the present study we show, by Western blotting and
electrophoretic mobility shift assay, that in human islets
of Langerhans incubated in low glucose (3 mM) PDX1
exists as an inactive 31-kDa protein localized exclu-
sively in the cytoplasm. Transfer of the islets to high (16
mM) glucose results in rapid (within 10 min) conversion
of PDX1 to an active 46-kDa form that was present pre-
dominantly in the nucleus. Activation of PDX1 appears
to involve phosphorylation, as shown by incorporation
of
32
P
i
into the 46-kDa form of the protein. These effects
of glucose could be mimicked by chemical stress (sodi-
um arsenite), or by overexpression of SAPK2 in the
b
-cell line MIN6. Overexpression of SAPK2 also stimu-
lated PDX1-dependent transcription of a –50 to –250 re-
gion of the human insulin gene promoter linked to a
firefly luciferase reporter gene. The effects of glucose
were inhibited by the SAPK2 inhibitor SB 203580, and by
wortmannin and LY 294002, which inhibit phosphatidyl-
inositol 3-kinase, although the effects of stress (arsenite)
were inhibited only by SB 203580. These results demon-
strate that glucose regulates the insulin gene promoter
through activation and nuclear translocation of PDX1
via the SAPK2 pathway.
The homeodomain transcription factor PDX1 (pancreatic/
duodenal homeobox-1), which has previously been called IPF1
(1), IDX1 (2), STF1 (3), or IUF1 (4), plays an important role in
lineage determination in the developing endocrine pancreas (5,
6). PDX1 binds to four sites (A1, A2, A3, and A5) with the
consensus sequence C(C/T)TAATG in the human insulin pro-
moter (7). Although it can transactivate the insulin promoter
(8–10), its exact role in basal insulin transcription is unclear
since insulin gene transcription can occur in the absence of
PDX1 (11, 12). However, there is strong evidence supporting a
role for PDX1 in the mechanisms whereby glucose stimulates
insulin gene transcription in response to changes in glucose
concentrations (13–15). Furthermore, glucose metabolism has
been shown to stimulate PDX1 DNA binding activity and in-
sulin promoter activity in
b
-cells via a stress-activated signal-
ing pathway involving stress-activated protein kinase 2
(SAPK2, also termed RK/p38, CSBP, and Mxi2) (16).
SAPK2 is a member of an expanding family of mitogen-
activated protein kinase-related kinases that are activated in
response to adverse stimuli such as heat, osmotic shock, ultra-
violet light, DNA-damaging reagents and by proinflammatory
cytokines that are produced under conditions of stress (17). In
pancreatic
b
-cells, glucose metabolism stimulates the SAPK2
pathway by a mechanism that involves phosphatidylinositol
3-kinase (PI 3-kinase),
1
while stress (e.g. arsenite treatment)
stimulates the SAPK2 pathway in
b
and other cell types by a
mechanism independent of PI 3-kinase (16).
Recombinant PDX1, synthesized in Escherichia coli, has a
molecular mass of 31 kDa, similar to that predicted from its
amino acid sequence. It is inactive, insofar as it does not bind to
its recognition sequence in the insulin promoter as measured
by electrophoretic mobility shift assay. However, treatment of
recombinant PDX1 with SAPK2 in the presence of Mg-ATP and
a
b
-cell extract activates its DNA binding, concomitant with a
shift in molecular mass from 31 kDa to 46 kDa (16). The
molecular modifications responsible for this shift in molecular
mass are as yet unclear. In the present study we show that
PDX1 exists as the inactive 31-kDa form in the cytoplasm of
human islets incubated in low (3 mM) glucose. Treatment with
16 mMglucose stimulates conversion to the active 46-kDa form,
which is localized predominantly in the nucleus. These events
are inhibited by SB 203580 (5
m
M) (an inhibitor of SAPK2),
wortmannin, and LY 294002 (inhibitors of PI 3-kinase), and
mimicked by sodium arsenite and overexpression of SAPK2.
EXPERIMENTAL PROCEDURES
Chemicals and Reagents—Radiochemicals were purchased from Am-
ersham International (Slough, Berks, United Kingdom (UK)), sodium
arsenite from Fisons (Loughborough, UK), and wortmannin from Sigma
(Poole, UK). SB 203580 was a generous gift from Dr. J. Lee and Dr. P.
Young (SmithKline Beecham, King of Prussia, PA). Anti-PDX1 anti-
body was a kind gift from Dr. C. V. Wright (Vanderbilt University,
Nashville, TN). Anti-SAPK2 antibody was purchased from New Eng-
land Biolabs (Cambridge, UK). Anti-upstream stimulatory factor (USF)
antibody was kindly provided by Dr. M. Sawadogo (University of Texas,
Houston, TX).
Oligonucleotides—Oligodeoxynucleotides were purchased from Alta
Bioscience (University of Birmingham, Birmingham, UK). Single-
stranded complementary oligonucleotides were annealed as described
* This work was supported by grants from the Wellcome Trust and
the British Diabetic Association and by a studentship (to C. M. M.) from
the British Diabetic Association. The costs of publication of this article
were defrayed in part by the payment of page charges. This article must
therefore be hereby marked “advertisement” in accordance with 18
U.S.C. Section 1734 solely to indicate this fact.
To whom correspondence should be addressed. Fax: 44-1224-
273144.
1
The abbreviations used are: PI 3-kinase, phosphatidylinositol 3-ki-
nase; USF, upstream stimulatory factor; EMSA, electrophoretic mobil-
ity shift assay; PAGE, polyacrylamide gel electrophoresis; IL, interleu-
kin; NF, nuclear factor.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 274, No. 2, Issue of January 8, pp. 1011–1016, 1999
© 1999 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
This paper is available on line at http://www.jbc.org 1011
by guest on November 15, 2016http://www.jbc.org/Downloaded from
previously (18), and labeled with [
g
-
32
P]ATP using T4 polynucleotide
kinase. Oligonucleotide B, corresponding to the A3 site of the human
insulin gene promoter, is a complementary double-stranded 30-mer
59-CCCCTGGTTAAGACTCTAATGACCCGCTGG-39.
Isolation and Treatment of Human Islets of Langerhans—Human
islets of Langerhans were isolated from the pancreas of human organ
donors, and all procedures were carried out with the approval of the
appropriate Ethical Committee. The pancreatic duct was cannulated in
situ and digestion achieved by intraductal infusion of collagenase. The
islets were then separated from contaminating acinar tissue by centrif-
ugation on discontinuous density gradients of bovine serum albumin in
a semi-automated system (19). The purified islets were placed in RPMI
1640 medium (Life Technologies, Inc.) containing 10% (v/v) fetal calf
serum and supplemented with 400 IU/ml sodium penicillin G and 200
m
g/ml streptomycin sulfate, and cultured at 37 °C in a humidified
atmosphere of O
2
:CO
2
(95:5) for several days prior to use. Selected islets
were separated into batches of 120–150 in Hanks’ buffered saline (0.12
MNaCl, 5.4 mMKCl, 0.33 mMNa
2
HPO
4
, 0.44 mMKH
2
PO
4
, 0.4 mM
MgSO
4
, 0.5 mMMgCl
2
,1mMCaCl
2
,2mMNaHCO
3
, 5% (w/v) bovine
serum albumin) except where specified.
Preparation of Cell Extracts—Nuclear and cytoplasmic extracts were
prepared using a modification of the method of Schreiber et al. (20).
Cells were centrifuged for 10 s in a microcentrifuge, and resuspended in
400
m
lof10mMHepes, pH 7.9, containing 10 mMKCl, 0.1 mMEDTA,
0.1 mMEGTA, 1 mMdithiothreitol, 0.5 mMphenylmethanesulfonyl
fluoride, 10 mMNaF, 10 mMsodium molybdate, 10 mM
b
-glycerophos-
phate, 10 mMsodium vanadate, and 10 mMp-nitrophenyl phosphate.
Cells were allowed to swell on ice for 15 min before adding 25
m
lof10%
(v/v) Nonidet P-40. The islets were then incubated for an additional 30
min on ice, vortexed for 15 s, and centrifuged for 30 s in a microcentri-
fuge. The cytoplasmic fraction (supernatant) was removed and frozen in
small aliquots, and the pellet, which was enriched in nuclei, was resus-
pended in 50
m
lof20mMHepes, pH 7.9, containing 0.4 MNaCl, 1 mM
EDTA, 1 mMEGTA, 1 mMdithiothreitol, 1 mMphenylmethanesulfonyl
fluoride, 10
m
g/ml leupeptin, 1
m
g/ml pepstatin A, 0.1 mMp-aminoben-
zoic acid, 10
m
g/ml aprotinin, 5% (v/v) glycerol, 10 mMNaF, 10 mM
sodium molybdate, 10 mM
b
-glycerophosphate, 10 mMsodium vanadate,
and 10 mMp-nitrophenyl phosphate. Nuclear extracts were then cen-
trifuged for 2 min at 4 °C in a microcentrifuge. The supernatant was
collected, aliquoted into small volumes, and stored at 270 °C. For whole
cell extracts, Nonidet P-40 lysis was replaced with incubation with 1%
(v/v) Triton X-100, vortexing for 1 min, and centrifugation for 2 min.
Electrophoretic Mobility Shift Assays (EMSAs)—EMSAs were per-
formed as described previously (16). Cell extracts (0.5
m
g of protein)
were incubated with radiolabeled probe for 20 min at room temperature
in buffer containing 10 mMTris-HCl, pH 7.5, 50 mMKCl, 5 mMdithio-
threitol, 1 mMEDTA, and 5% (v/v) glycerol.
Cell Culture—MIN6 cells were grown in Dulbecco’s modified Eagle’s
medium containing 5 mMglucose, supplemented with 15% heat-inacti-
vated myoclone fetal calf serum (Sigma) and 2 mML-glutamine, in a
humidified atmosphere containing 95% air, 5% CO
2
.
Plasmids—The control construct pGL-LUC is based on the plasmid
pGL2 (Promega), with the thymidine kinase promoter from the herpes
simplex virus cloned 59to the firefly luciferase gene. The construct
pGL-LUC200 varies from this in that it contains 200 base pairs of the
human insulin gene promotor. A 260 to 2260 base pair HincII-PvuII
fragment from the human insulin gene promoter was blunt-ended and
cloned into the SmaI site of the control construct. DNA was prepared
using the Qiagen Endotoxin-Free Maxiprep method, and quantitated
spectrophotometrically. PDX1 expression vector pCR3-PDX1 was as
described previously (11). The pSG5-SAPK2 (21) expression vector was
a generous gift from Dr. S. Keyse, University of Dundee.
Transfections—Cells were grown to about 80% confluence in six-well
plates, and were transfected by mixing 4
m
g of DNA and 54
m
lofa1nM
lipid suspension containing a 2:1 mixture of dioleoyl-L-
a
-phosphati-
dylethanolamine (Sigma) and dimethyl-dioctadecylammonium bromide
(Fluka) in 1 ml of serum free Opti-MEM (Life Technologies, Inc.). The
lipid-DNA complexes were allowed to form for 20 min at room temper-
ature before being added to the washed cells. Following5hofincuba-
tion, 1 ml of complete medium containing 30% heat-inactivated
myoclone fetal calf serum was added to the cells. After 12 h, the
medium-DNA complexes were replaced by complete medium and the
cells left for another 24 h before treating. Treatment of all cells started
with a 5-h preincubation in Dulbecco’s modified Eagle’s medium con-
taining 0.5 mMglucose. Cells were then removed from the wells and a
cell pellet recovered by centrifugation at 7000 rpm for 30 s. The cell
pellet was resuspended in 70
m
l of 100 mMKH
2
PO
4
,pH7.8,1mM
dithiothreitol solution and lysed by freeze/thawing three times. Cell
debris was removed by centrifugation at 13,000 rpm for 1 min.
Luciferase Assay—30
m
l of cell extract was added to 350
m
l of buffer
A, pH 7.8 (15 mMMgSO
4
,30mMglycylglycine, 2 mMNa
2
ATP) contain-
ing 0.45 mMcoenzyme A and 2.56 mMTriton X-100. To this, 150
m
lof
buffer G (30 mMglycylglycine) containing 0.5 mMluciferin (Sigma) was
injected and the luminescence read at 560 nMusing a Berthold Luma
LB9501. Protein content of the cell extract was measured by taking 6
m
l
of extract and adding144
m
l of water and 150
m
l of Coomassie protein
assay reagent (Pierce). Protein content was read at a wavelength of 620
nm using a Titer Tech Multiscan MCC340. A standard curve of known
bovine serum albumin protein concentrations was used to calculate the
protein concentrations of the samples.
Western Blotting—For Western blot analysis, 1-
m
g samples of cell
extract were fractionated by SDS-PAGE, blotted on to ECL-nitrocellu-
lose membrane (Amersham), and incubated for 60 min in a buffer
containing 10 mMTris-HCl, 0.05% (v/v) Tween 20, 0.5 MNaCl, and a
1:5000 dilution of anti-PDX1 antibody. The antigen-antibody complex
was then detected by incubating the membrane for another 60 min in
buffer containing a 1:5000 dilution of horseradish peroxidase-conju-
gated anti-rabbit IgG secondary antibody (ECL, Amersham).
32
P
i
Labeling and PDX1 Immunoprecipitation—MIN6 cells were
washed three times in Krebs Ringer (118 mMNaCl, 4.75 mMKCl, 1.2
mMMgCl
2
, 0.26 mMCaCl
2
,25mMNaHCO
3
) prior to addition of 0.5 mCi
of
32
P
i
/10
6
cells in Krebs Ringer. Cells were incubated at 37 °C for 1 h,
prior to addition of glucose to 0.5 or 16 mMfinal concentration. After 30
min cells were washed three times in Krebs Ringer and harvested, with
nuclear and cytoplasmic extracts prepared as before. For PDX1 immu-
noprecipitation: 400-
m
l samples in 13NDET (1% Nonidet P-40, 0.4%
deoxycholate, 66 mMEDTA, 10 mMTris-HCl, pH 7.4) were precleared
by addition of 5
m
l of preimmune rabbit serum and 10
m
l of protein A (in
13NDET), with rotation at 4 °C for 2 h. Preclearing was completed by
centrifugation at 13,000 rpm for 2 min at 4 °C, with the resulting pellet
being discarded. 3
m
l of anti-PDX1 antibody and 10
m
l of protein A-
Sepharose were added to 400
m
l of supernatant, and PDX1 immuno-
precipitated by rotation at 4 °C for 16 h. A 2-min centrifugation yielded
a pellet, which was then resuspended in 0.5 ml of NDET 10.3% SDS,
with rotation at 4 °C for 2 h. Samples were then layered over 0.5 ml of
NDET 10.3% SDS/30% sucrose, and centrifuged for 5 min at 4 °C. The
resulting pellet was resuspended in 0.5 ml of NDET and rotated at 4 °C
for another 2 h. Finally, the samples were centrifuged for 4 min at 4 °C,
with the final pellet resuspended in 30
m
l of SDS-PAGE sample buffer
(4% SDS, 0.02% bromphenol blue, 0.5 Msucrose, 10 mMTris-HCl, pH
6.8, 10%
b
-mercaptoethanol). Samples were heated to 80 °C for 5 min
prior to loading.
RESULTS
Initial experiments were undertaken to determine the effect
of glucose on the molecular mass and intracellular location of
PDX1. Human islets of Langerhans were incubated for5hin3
mMglucose, transferred to 16 mMglucose, and PDX1 analyzed
at various time intervals by Western blotting of whole cell
extracts. In 3 mMglucose PDX1 was present as a 31-kDa
protein (Fig. 1A). Within 10 min in 16 mMglucose, the 31-kDa
protein was converted to the 46-kDa form. Conversion was
complete within 15 min. To investigate the effect of glucose on
the intracellular location of the two forms of PDX1, human
islets were incubated in 3 or 20 mMglucose and cytoplasmic
and nuclear fractions prepared. In 3 mMglucose, the 31-kDa
form was present in the cytoplasm; however, in 16 mMglucose,
the 46-kDa form was present in the cytoplasm and the nucleus
(Fig. 1B). Treatment with acid phosphatase converted the 46-
kDa form to the 31-kDa form. In the presence of excess inor-
ganic pyrophosphate, acid phosphatase had no effect on the
46-kDa form, confirming that conversion to the 31-kDa form
was associated with dephosphorylation of the 46-kDa form. The
31-kDa form was inactive as measured by EMSA (Fig. 1C,
lanes 1–4), whereas the 46-kDa form present in the cytoplasm
and the nucleus was active (Fig. 1C,lanes 48). Identification
of the retarded band in Fig. 1Cas PDX1 was confirmed by
oligonucleotide and antibody competition experiments (data
not shown).
The role of phosphorylation in the activation of PDX1 was
investigated further by labeling of MIN6 cells with inorganic
Nuclear Translocation of PDX11012
by guest on November 15, 2016http://www.jbc.org/Downloaded from
32
P
i
and subsequent immunoprecipitation of PDX1. At high
glucose concentrations,
32
P
i
was incorporated into the 46-kDa
form of PDX1 (Fig. 2, lanes 3 and 4), but at low glucose con-
centrations no such incorporation was observed (lanes 1 and 2).
Western blot analysis of the immunoprecipitated samples (Fig.
2B) confirmed that both forms of the protein were present, but
that
32
P was incorporated only into the 46-kDa form of the
protein, which was observed at high glucose concentrations. To
evaluate the effectiveness of the biochemical fractionation of
the
b
-cell extracts, nuclear and cytoplasmic fractions were an-
alyzed by Western blotting using a specific anti-USF antibody.
USF binds to the E2 site of the human insulin gene promoter
and has a molecular size of 43 kDa (Fig. 2C). Blotting con-
firmed the presence of USF solely in the nuclear fractions,
independently of glucose concentrations.
Since SAPK2 and PI 3-kinase had previously been impli-
cated in the stimulation of PDX1 DNA binding activity, the
possible involvement of these enzymes in the glucose-stimu-
lated translocation of PDX1 to the nucleus was next investi-
gated. Fig. 3Ashows that glucose-stimulated conversion of
PDX1 from the 31-kDa to the 46-kDa form and translocation of
the 46-kDa form into the nucleus was inhibited by the SAPK2
inhibitor SB 203580, and by the PI 3-kinase inhibitors wort-
mannin and LY 294002 (Fig. 3A). SAPK2 is a stress-activated
protein kinase, the activity of which can be stimulated by
treating cells with the stress-inducing agent sodium arsenite (1
mM). The effects of glucose on PDX1 molecular mass and intra-
cellular location were mimicked by sodium arsenite, further
supporting a role for SAPK2 in these events (Fig. 3B). As shown
previously (16) the effects of arsenite were inhibited by SB
203580 but not by wortmannin and LY 294002 (Fig. 3B). The
IC
50
for SB 203580 on both glucose and arsenite effects was 3
m
M(data not shown).
To investigate further the involvement of SAPK2, MIN6 cells
were transfected with a cDNA encoding SAPK2. Overexpres-
sion was confirmed by the appearance of a 38-kDa immunore-
active protein in extracts from transfected cells that was absent
in untransfected cells (Fig. 4A). In untransfected MIN6 cells
treated with 3 mMglucose, PDX1 was present as the 31-kDa
form in the cytoplasm. Following treatment with 16 mMglu-
cose, PDX1 was converted to the 46-kDa form, which was
present in the cytoplasm and nucleus. When cells transfected
with SAPK2 were incubated in 3 mMglucose, PDX1 was pres-
ent as the 46-kDa form in the cytoplasm and in the nucleus
(Fig. 4B). This result suggested that overexpression of SAPK2
resulted in conversion of PDX1 to the 46-kDa form that was
localized predominantly in the nucleus. Overexpression of
SAPK2 also activated PDX1 DNA binding activity in low glu-
cose-treated MIN6 cells (Fig. 4C). The critical events governing
modification and activation of PDX1 appear to occur in the
cytoplasm, as Western blot analysis of nuclear and cytoplasmic
extracts prepared from MIN6 cells overexpressing SAPK2
clearly shows that the SAPK2 protein is localized in the cyto-
plasm and is undetectable in the
b
-cell nucleus (Fig. 5A).
As shown previously (16), glucose stimulates the transcrip-
tional activity of a DNA construct containing the firefly lucif-
erase gene under the control of the thymidine kinase reporter,
which is driven by a human insulin gene fragment from 260 to
2260 (pGL -LUC200) (Fig. 5). Overexpression of SAPK2 in
MIN6 cells had no effect on the control vector pGL-LUC, which
lacked the insulin gene fragment, but gave a 5-fold stimulation
of pGL-LUC200 activity, similar to that seen with 16 mMglu-
cose (Fig. 5). The effect of overexpression of SAPK2 on the
LUC200 activity was inhibited by SB 203580 (10
m
M), confirm-
ing that these effects were the direct consequence of the higher
SAPK2 activity in the transfected cells.
DISCUSSION
We have previously shown that glucose activates PDX1 DNA
binding activity and insulin promoter activity in pancreatic
b
-cells via a pathway involving SAPK2 (RK/p38). We also
showed that SAPK2 in vitro activated recombinant PDX1 DNA
binding activity, concomitant with a change in the molecular
mass of PDX1 from 31 kDa to 46 kDa (16). In the present study,
we show that glucose stimulates the conversion of PDX1 from
a 31-kDa unphosphorylated to a 46-kDa phosphorylated form
in isolated human islets of Langerhans (Fig. 1) and in MIN6
cells (Fig. 4). The activation of PDX1, and translocation to the
nucleus, were inhibited by SB 203580 and stimulated by ar-
senite and overexpression of SAPK2. This strongly supports a
role for SAPK2 in these events. That glucose stimulates a rapid
translocation of PDX1 to the nucleus is compatible with the
results of a recent study showing that glucose stimulates trans-
location of a c-Myc-tagged PDX1 (IPF-1) from the nuclear pe-
riphery to the nucleoplasm (22).
SAPK2-dependent phosphorylation of PDX1 appears to rep-
FIG.1.PDX1 is rapidly modified by
high glucose concentrations in hu-
man islets of Langerhans. A, Western
blot analysis of human islets of Langer-
hans incubated in 3 mMglucose for 5 h,
then transferred to 20 mMglucose for the
time periods indicated. Whole cell ex-
tracts were prepared, and probed with a
specific anti-PDX1 antibody. B, Western
blot analysis of cytoplasmic (C) and nu-
clear (N) extracts prepared from human
islets of Langerhans incubated in 3 mM
glucose for 5 h, or 3 mMglucose for 5 h
followed by 20 mMglucose for 30 min. In
lane 5, high glucose nuclear extract was
incubated for 30 min with 10 units of po-
tato acid phosphatase (AP). In lane 6, acid
phosphatase treatment was carried out in
the presence of 50 mMpyrophosphate (P
i
).
C, electrophoretic mobility shift assay of
cytoplasmic (C) and nuclear (N) samples
prepared from human islets of Langer-
hans incubated in 3 mMglucose for 5 h, or
3m
Mglucose for 5 h followed by 20 mM
glucose for 30 min, using the A3 site of the
human insulin gene promoter as a probe.
Nuclear Translocation of PDX1 1013
by guest on November 15, 2016http://www.jbc.org/Downloaded from
resent the critical step in glucose-induced translocation to the
nucleus and activation of DNA binding activity. However, it is
unclear whether phosphorylation alone accounts for the ob-
served change in size. It is possible that phosphorylation allows
or promotes a second event, which is yet to be characterized.
Phosphorylation may induce a conformational change in PDX1
that affects its mobility in SDS-PAGE, but the change in size
could equally result from a further post-translational modifi-
cation. Many transcription factors contain nuclear localization
signals within their amino acid sequences, but others, such as
PDX1, do not contain recognizable nuclear localization signals
and may require modification at the post-translational level.
FIG.2. High glucose promotes in-
corporation of
32
P
i
into the 46-kDa
form of PDX1. A, SDS-PAGE analysis of
PDX1 immunoprecipitated from nuclear
(N) and cytoplasmic (C) extracts prepared
from MIN6 cells incubated for3hin0.5
mMglucose, then labeled with
32
P
i
. La-
beled cells were incubated in 0.5 mMglu-
cose (lanes 1 and 2)or16m
Mglucose
(lanes 3 and 4) for 30 min prior to the
preparation of extracts.
32
P-Labeled
PDX1 is observed in lanes 3 and 4at 46
kDa. B, Western blot analysis of immuno-
precipitates described in A, using a spe-
cific anti-PDX1 antibody. C, Western blot
analysis of the cytoplasmic (C) and nu-
clear (N) extracts from MIN6 cells incu-
bated in 0.5 mMglucose for 5 h, or incu-
bated in 0.5 mMglucose for 5 h followed by
stimulation in 16 mMglucose for 30 min,
using a specific anti-USF antibody. USF
has a molecular mass of 43 kDa.
FIG.3. High glucose and chemical
stress induce modification of PDX1.
A, Western blot (anti-PDX1) analysis of
cytoplasmic (C,lanes 3–6), and nuclear
(N,lanes 7–10) samples prepared from
human islets of Langerhans incubated in
3m
Mglucose for5h(lanes 1 and 2), or 3
mMglucose for 5 h followed by 20 mM
glucose for 30 min, or in 20 mMglucose for
30 min in the presence of the inhibitors
indicated. SB 203580 (SB,20
m
M)LY
294002 (LY,50
m
M), and wortmannin
(Wtm,50n
M) were added 30 min prior to
stimulation. B, Western blot (anti-PDX1)
analysis of cytoplasmic (C,lanes 3–6),
and nuclear (N,lanes 7–10) samples pre-
pared from human islets of Langerhans
incubated in 3 mMglucose for5h(lanes 1
and 2), or in 3 mMglucose for 5 h followed
by1m
Msodium arsenite for 30 min in the
presence of the inhibitors indicated.
SB203580 (SB,20
m
M) LY294002 (LY,50
m
M), and wortmannin (Wtm,50nM) were
added 30 min prior to stimulation.
Nuclear Translocation of PDX11014
by guest on November 15, 2016http://www.jbc.org/Downloaded from
Common modifications promoting nuclear localization include
glycosylation (23), and ubiquitination (24, 25), which modifies
proteins through the addition of ubiquitin-like protein species
such as SUMO1 (small ubiquitin-related modifier 1). In the
case of RanGAP (Ran-GTPase activating protein) (25), phos-
phorylation allows the addition of SUMO1, which alters the
apparent molecular mass of the protein by 20 kDa, and pro-
motes its translocation from the cytoplasm to the nucleus.
Similar events may be occurring with PDX1.
Increased binding activity of transcription factors through
phosphorylation can occur by several mechanisms. Many tran-
scription factors become phosphorylated in the cytoplasm,
stimulating movement into the nucleus (26). However, this is
not always the case. For example, insulin stimulates the nu-
clear translocation of several kinases, such as mitogen-acti-
vated protein kinase (27), which can then directly phosphoryl-
ate nuclear protein substrates, or activate other kinases (28).
In the present study SAPK2 is shown to be present exclusively
in the cytoplasm, suggesting that the events governing PDX1
activation occur exclusively in the cytoplasm, promoting trans-
location of an active PDX1 into the nucleus.
Glucose activation of PDX1 nuclear translocation occurs
through a
b
-cell signaling pathway involving PI 3-kinase, and
resulting in the activation of SAPK2. Hence, overexpression of
SAPK2 can artificially promote PDX1 translocation to the nu-
cleus even in lower glucose concentrations. In fact, overexpres-
sion of SAPK2 stimulated PDX1 binding activity to levels
higher than those seen in high glucose (Fig. 4). This hyper-
stimulation of PDX1 by overexpression of SAPK2 occurred in
0.5 or 20 mMglucose, and appeared to reflect activation of
PDX1 at maximum efficiency. In untransfected
b
-cells endog-
enous SAPK2 may represent a rate-limiting step in PDX1
activation, with glucose eliciting a controlled stimulation.
Overexpression of SAPK2 to artificially high levels may there-
fore result in a hyperstimulation of the PDX1 activation path-
way, resulting in an increased ability to phosphorylate and
translocate all available PDX1 protein. This “hyperstimula-
tion” of PDX1 binding activity did not appear to be reflected in
the levels of insulin gene transcription relative to those ob-
served in high glucose. This is not surprising, as PDX1 alone
does not control transcription of the insulin gene. Several other
factors (e.g. E1 and E2 site binding factors) are absolutely
required for transcription of the insulin gene. Increased PDX1
binding activity will only exert an effect in the presence of these
factors, and a maximum response would require their com-
bined activities.
Glucose stimulates PDX1 phosphorylation and nuclear
translocation through SAPK2. PDX1 is not unique in this re-
spect; CREB, which also binds to the human insulin gene
promoter (15, 16), has its binding activity controlled through
alteration in its phosphorylation status. Recent studies in SK-
N-MC cells have shown that CREB-dependent transcription is
triggered by co-transfection with SAPK2 (29), which may act in
this case through stimulation of a downstream kinase such as
MAPKAPK2 (30). However, SAPK2 has been shown to directly
phosphorylate several transcription factors, including ELK1,
CHOP, and MEF2C (17). Stress-activated protein kinases have
also been implicated in the nuclear localization of other tran-
scription factors. In EL-4 D6/76 cells, IL-1 activation of inter-
leukin-1 receptor-associated kinase and of stress-activated pro-
FIG.4.Overexpression of SAPK2 mimics the effect of high glucose on PDX1. A, Western blot analysis of untransfected MIN6 cell extract
(lanes 1–3), and extract prepared from MIN6 cells transfected with pSG-SAPK2 (lanes 4–6), using a specific anti-SAPK2 antibody. SAPK2 has an
apparent molecular mass of 38 kDa. B, Western blot analysis (anti-PDX1) of cytoplasmic (lanes 1–4) or nuclear (lanes 5–8) extracts prepared from
untransfected MIN6 cells (2), or MIN6 cells overexpressing SAPK2 (1). Samples were incubated in 0.5 mMglucose for 5 h, or 0.5 mMglucose for
5 h followed by stimulation for 30 min at 16 mMglucose, as indicated. C, electrophoretic mobility shift assay of MIN6 nuclear extracts prepared
from cells incubated in 0.5 mMglucose for 5 h (lanes 1–3), 0.5 mMglucose for 5 h followed by 30 min at 16 mMglucose (lanes 46), or cells
overexpressing SAPK2 which were incubated in 0.5 mMglucose for 5 h. EMSA analysis was performed using the A3 site of the human insulin gene
promoter as probe.
Nuclear Translocation of PDX1 1015
by guest on November 15, 2016http://www.jbc.org/Downloaded from
tein kinases, promotes translocation of transcription factors
NF-
k
B and IL-1 NF to the nucleus, resulting in the induction of
IL-2 mRNA synthesis (30). The phosphorylation induced nu-
clear translocation of Rel family proteins like NF-
k
B is induced
by an extraordinary large number of agents (26), cellular stress
being one example from many.
It has been proposed that glucose increases endogenous in-
sulin mRNA levels and insulin promoter activity in HIT T15
cells via exocytosis of insulin leading to autocrine stimulation
of the
b
-cell insulin receptor (31). The insulin stimulatory ef-
fects on mRNA levels were mediated through PI 3-kinase, p70
S6 kinase, and calmodulin kinase pathways. We have also
found that insulin (1–10 ng/ml) stimulates PDX1 DNA binding
activity in human islets of Langerhans and the human insulin
promoter (pGL-LUC200) activity in MIN-6 cells.
2
However, in
our study the effects of insulin were inhibited by the PI 3-ki-
nase inhibitors wortmannin (50 nM) and LY 294002 (10
m
M) and
by the SAPK2 inhibitor SB 203580 (7
m
M), but not by the p70 S6
kinase inhibitor rapamycin (50
m
M). This suggests that modu-
lation of endogenous insulin mRNA levels by insulin may in-
volve additional pathways to those described for the activation
of PDX1.
In summary, glucose stimulates translocation of the home-
odomain transcription factor PDX1 from the cytoplasm to the
nucleus in pancreatic
b
-cells via a cell signaling pathway in-
volving SAPK2. This translocation is associated with a 5-fold
increase in insulin promoter activity, and may represent a
pivotal event in the nutrient regulation of insulin mRNA
production.
Acknowledgment—We thank Prof. P. Cohen for helpful suggestions
and provision of reagents.
REFERENCES
1. Ohlsson, H., Karlsson, K., and Edlund, T. (1993) EMBO J. 12, 4251–4259
2. Miller, C. P., McGehee, R. E., Jr., and Habener, J. F. (1993) EMBO J. 13,
1145–1156
3. Leonard, J., Peers, B., Johnson, T., Ferreri, S. L., and Montminy, M. R. (1993)
Mol. Endocrinol. 7, 1275–1283
4. Boam, D. W. S., and Docherty, K. (1989) Biochem. J. 264, 233–239
5. Jonsson, J., Carlsson, L., Edlund, T., and Edlund, H. (1994) Nature 371,
606–609
6. Guz, Y., Montminy, M. R., Stein, R., Leonard, J., Gamer, L. W., Wright,
C. V. E., and Teitelman, G. (1995) Development 121, 11–18
7. Clark, A. R., Petersen, H., Read, M. L., Scott, V., Michelsen, B., and Docherty,
K. (1993) FEBS Lett. 329, 139–143
8. Petersen, H. V., Serup, P., Leonard, J., Michelsen, B. K., and Madsen, O. D.
(1994) Proc. Natl. Acad. Sci. U. S. A. 91, 10465–10469
9. Peers, B. J., Leonard, J., Sharma, S., Teitelman, G., and Montminy, M. R.
(1995) Mol. Endocrinol. 8, 1798–1806
10. Serup, P., Jensen, J., Andersen, F. G., Jorgensen, M. C., Blume, N., Holst, J. J.,
and Madsen, O. D. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 9015–9020
11. Macfarlane, W. M., Cragg, H., Docherty, H. M., Read, M. L., James, R. F. L.,
Aynsley-Green, A., and Docherty, K. (1997) FEBS Lett. 413, 304–308
12. Kajimoto, Y., Watabe, H., Matsuoka, T., Kaneto, H., Fujitani, Y., Miyazaki,
J.-I., and Yamasaki, Y. (1997) J. Clin. Invest. 100, 1840–1846
13. Melloul, D., Ben-Neriah., Y., and Cerasi, E. (1993) Proc. Natl. Acad. Sci.
U. S. A. 90, 3865–3869
14. Macfarlane, W. M., Read, M. L., Gilligan, M., Bujalska, I., and Docherty, K.
(1994) Biochem. J. 303, 625–631
15. Marshak, S., Totary, H., Cerasi, E., and Melloul, D. (1996) Proc. Natl. Acad.
Sci. U. S. A. 93, 15057–15062
16. Macfarlane, W. M., Smith, S. B., James, R. F. L., Clifton, A. D., Doza, Y. N.,
Cohen, P., and Docherty, K. (1997) J. Biol. Chem. 272, 20936–20944
17. Cohen, P. (1997) Trends Cell Biol. 7, 353–361
18. Boam, D. S., Clark, A. R., and Docherty, K., (1990) J. Biol. Chem. 265,
8285–8296
19. Ricordi, C., Lacy, P. E., and Scharp, D. W. (1989) Diabetes 38, 140–145
20. Schreiber, E., Matthias, P., Muller, M. M., and Schaffner, W. (1989) Nucleic
Acids Res. 17, 6419
21. Groom, L. A., Sneddon, A. A., Alessi, D. R., Dowd, S., and Keyse, S. M. (1996)
EMBO J. 15, 3621–3632
22. Rafiq, I., Kennedy, H. J., and Rutter, G. A. (1998) J. Biol. Chem. 273,
23241–23247
23. Duverger, E., Pellerin-Mendes, C., Mayer, R., Roche, A., and Monsigny, M
(1995) J. Clin. Sci. 108, 1325–1332
24. Mahajan, R., Delphin, C., Guan, T., Gerace, L., and Melchiar, F (1997) Cell 88,
97–107
25. Matunis, M. J., Coutavas, E., and Blobel, G (1996) J. Cell Biol. 135, 1457–1470
26. Thanos, D., and Maniatis, T (1995) Cell 80, 529–532
27. Chen, R., Sarnecki, C., and Blenis, J. (1992) Mol. Cell. Biol. 12, 915–927
28. Traverse, S., Gomez, N., Paterson, H., Marshall, C., and Cohen, P. (1992)
Biochem. J. 288, 351–355
29. Kim, J., and Kahn, C. R. (1997) Biochem. J. 323, 621–627
30. Tan, Y., Rouse, J., Zhang, A., Cariati, S., Cohen, P., and Comb, M. J. (1996)
EMBO J. 15, 4629–4642
31. Leibiger, I. B., Leibiger, B., Moele, T, and Berggren, P.-O. (1998) Mol. Cell 1,
933–938
2
H. Wu, W. M. Macfarlane, and K. Docherty, unpublished results.
FIG.5. Overexpression of SAPK2 mimics the effect of high
glucose concentrations on pGL-Luc200 reporter gene activity.
A, Western blot analysis of cytoplasmic (C) and nuclear (N) extracts
prepared from MIN6 cells overexpressing SAPK2, using a specific anti-
SAPK2 antibody. SAPK2 has an apparent molecular mass of 38 kDa. B,
MIN6 cells were transfected with the control construct pGL-Luc (LUC,
lanes 1–4), or with pGL-Luc200 (LUC200,lanes 5–8), and were incu-
bated in 0.5 mM(white)or16mMglucose (black), or 0.5 mMglucose in
the presence of 20
m
MSB 203580 (hatched). In lanes 3,4, and 7–9, cells
were co-transfected with SAPK2 as indicated.
Nuclear Translocation of PDX11016
by guest on November 15, 2016http://www.jbc.org/Downloaded from
Roger F. L. James and Kevin Docherty
Wendy M. Macfarlane, Caroline M. McKinnon, Zoe A. Felton-Edkins, Helen Cragg,
-Cellsβfrom the Cytoplasm to the Nucleus in Pancreatic
Glucose Stimulates Translocation of the Homeodomain Transcription Factor PDX1
doi: 10.1074/jbc.274.2.1011
1999, 274:1011-1016.J. Biol. Chem.
http://www.jbc.org/content/274/2/1011Access the most updated version of this article at
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/274/2/1011.full.html#ref-list-1
This article cites 31 references, 16 of which can be accessed free at
by guest on November 15, 2016http://www.jbc.org/Downloaded from
... We show that both Pdx1 gene expression and protein levels were increased in MIN6B1 cells exposed to IMT504 for 48 h, indicating that IMT504 favors the maintenance of βcell identity. One molecular mechanism of action through which chronic hyperglycemia can cause worsening βcell function is via decreased protein expression of Pdx1 (Robertson, 2004), whereas the positive effects of glucose on insulin transcription may be a result of its enhancement of Pdx1 nuclear translocation (Macfarlane et al., 1999). Based on our results, we postulate that IMT504 may protect βcells exposed to high glucose levels during diabetes development, as suggested by our in vivo experiments. ...
Article
Full-text available
We have demonstrated that oligodeoxynucleotide IMT504 promotes significant improvement in the diabetic condition in diverse animal models. Based on these results, here we evaluated whether these effects observed in vivo could be due to direct effects on β-cells. We demonstrate by immunofluorescence that IMT504 enters the cell and locates in cytoplasm where it induces GSK-3β phosphorylation that inactivates this kinase. As GSK-3β tags Pdx1 for proteasomal degradation, by inactivating GSK-3β, IMT504 induces an increase in Pdx1 protein levels, demonstrated by Western blotting. Concomitantly, an increase in Ins2 and Pdx1 gene transcription was observed, with no significant increase in insulin content or secretion. Enhanced Pdx1 is promising since it is a key transcription factor for insulin synthesis and is also described as an essential factor for the maintenance β-cell phenotype and function. Dose-dependent inhibition of H2 O2 -induced apoptosis determined by ELISA as well as decreased expression of Bax was also observed. These results were confirmed in another β-cell line, beta-TC-6 cells, in which a cytokine mix induced apoptosis that was reversed by IMT504. In addition, an inhibitor of IMT504 entrance into cells abrogated the effect IMT504. Based on these results we conclude that the β-cell recovery observed in vivo may include direct effects of IMT504 on β-cells, by maintaining their identity/phenotype and protecting them from oxidative stress and cytokine-induced apoptosis. Thus, this work positions IMT504 as a promising option in the framework of the search of new therapies for type I diabetes treatment.
... Homeobox transcription factors are involved in spatial patterning during embryogenesis and have been documented to be dysregulated in cancer progression (Abate-Shen, 2002). Of particular interest is PBX1, which is a proposed pioneer factor with dysregulation connected to cancer progression, stimulated by glucose, and has direct binding to NMIIs (Macfarlane et al., 1999;Huang et al., 2003;Grebbin and Schulte, 2017). However, PBX1 was altered only in S1935D (log 2 (fold change) increase = 1.36)-and ...
Article
Nonmuscle myosin IIB (NMIIB) is considered a primary force generator during cell motility. Yet, many cell types, including motile cells, do not necessarily express NMIIB. Given the potential of cell engineering for the next wave of technologies, adding back NMIIB could be a strategy for creating super-cells with strategically altered cell morphology and motility. However, we wondered what unforeseen consequences could arise from such an approach. Here, we leveraged pancreatic cancer cells, which do not express NMIIB. We generated a series of cells where we added back NMIIB and strategic mutants that increase the ADP-bound time or alter the phosphorylation control of bipolar filament assembly. We characterized the cellular phenotypes and conducted RNA-seq analysis. The addition of NMIIB and the different mutants each has specific consequences for cell morphology, metabolism, cortical tension, mechanoresponsiveness, and gene expression. Major modes of ATP production are shifted, including alterations in spare respiratory capacity and the dependence upon glycolysis or oxidative phosphorylation. Several metabolic and growth pathways undergo significant changes in gene expression. This work demonstrates that NMIIB is highly integrated with many cellular systems and simple cell engineering has profound impact that extends beyond the primary contractile activity presumably being added to the cells.
... PDX-1 mainly exists in the cytoplasm or around the nucleus in the resting state. Changes in the external environment, such as ionizing radiation and the increase in glucose concentration, can activate PDX-1 and translocate it into the nucleus [50]. The NLS of transcription factors is a crucial requirement for its action, possibly because free PDX-1 is modified by phosphorylation, acetylation, and sumoylation, which exposes the NLS and guides the nuclear translocation of PDX-1 [51][52][53]. ...
Article
Full-text available
The pancreatic duodenum homeobox-1 (PDX-1) is a transcription factor encoded by a Hox-like homeodomain gene that plays a crucial role in pancreatic development, β-cell differentiation, and the maintenance of mature β-cell functions. Research on the relationship between PDX-1 and diabetes has gained much attention because of the increasing prevalence of diabetes melitus (DM). Recent studies have shown that the overexpression of PDX-1 regulates pancreatic development and promotes β-cell differentiation and insulin secretion. It also plays a vital role in cell remodeling, gene editing, and drug development. Conversely, the absence of PDX-1 increases susceptibility to DM. Therefore, in this review, we summarized the role of PDX-1 in pancreatic development and the pathogenesis of DM. A better understanding of PDX-1 will deepen our knowledge of the pathophysiology of DM and provide a scientific basis for exploring PDX-1 as a potential target for treating diabetes.
... Both transcription factors have been found to have a dual role, both in the initial steps of pancreatic progenitor specification and in the cell type's differentiation. In particular, Ptf1a is known as essential for the differentiation of acinar cells (Zecchin et al., 2004), while Pdx1 is reutilized later in development to promote specifically β-cells differentiation (Macfarlane et al., 1999;Afelik et al., 2006). The tightly modulated serial activation of different transcription factor regulatory modules will give rise to distinct pancreatic types. ...
Article
Full-text available
The identity and function of a given cell type relies on the differential expression of gene batteries that promote diverse phenotypes and functional specificities. Therefore, the identification of the molecular and morphological fingerprints of cell types across taxa is essential for untangling their evolution. Here we use a multidisciplinary approach to identify the molecular and morphological features of an exocrine, pancreas-like cell type harbored within the sea urchin larval gut. Using single cell transcriptomics, we identify various cell populations with a pancreatic-like molecular fingerprint that are enriched within the S. purpuratus larva digestive tract. Among these, in the region where they reside, the midgut/stomach domain, we find that populations of exocrine pancreas-like cells have a unique regulatory wiring distinct from the rest the of the cell types of the same region. Furthermore, Serial Block-face scanning Electron Microscopy (SBEM) of the exocrine cells shows that this reported molecular diversity is associated to distinct morphological features that reflect the physiological and functional properties of this cell type. Therefore, we propose that these sea urchin exocrine cells are homologous to the well-known mammalian pancreatic acinar cells and thus we trace the origin of this particular cell type to the time of deuterostome diversification. Overall, our approach allows a thorough characterization of a complex cell type and shows how both the transcriptomic and morphological information contribute to disentangling the evolution of cell types and organs such as the pancreatic cells and pancreas.
... observed in beta cells has been described in brain neurons by Ishikawa et al. 3 . The metabolic microenvironment could influence the localization of the protein, as has been observed of the beta-cell transcription factor Pdx1 26,27 . The RNAseq data show decreased expression of H3f3b, encoding H3.3. ...
Article
Full-text available
We identified two NEXMIF variants in two unrelated individuals with non-autoimmune diabetes and autistic traits, and investigated the expression of Nexmif in mouse and human pancreas and its function in pancreatic beta cells in vitro and in vivo. In insulin-secreting INS-1E cells, Nexmif expression increased strongly in response to oxidative stress. CRISPR Cas9-generated Nexmif knockout mice exhibited a reduced number of proliferating beta cells in pancreatic islets. RNA sequencing of pancreatic islets showed that the downregulated genes in Nexmif mutant islets are involved in stress response and the deposition of epigenetic marks. They include H3f3b, encoding histone H3.3, which is associated with the regulation of beta-cell proliferation and maintains genomic integrity by silencing transposable elements, particularly LINE1 elements. LINE1 activity has been associated with autism and neurodevelopmental disorders in which patients share characteristics with NEXMIF patients, and can cause genomic instability and genetic variation through retrotransposition. Nexmif knockout mice exhibited various other phenotypes. Mortality and phenotypic abnormalities increased in each generation in both Nexmif mutant and non-mutant littermates. In Nexmif mutant mice, LINE1 element expression was upregulated in the pancreas, brain, and testis, possibly inducing genomic instability in Nexmif mutant mice and causing phenotypic variability in their progeny.
Chapter
Since the discovery of nitric oxide (NO) as an important mediator of vasoregulation, the molecule has been found to be involved in several physiological and pathological processes in human. One area of recent interest is the potential role of NO in modulation of insulin secretion. Emerging data suggest that NO augments insulin release from pancreatic beta cells through increasing intracellular Ca2+ level or via S-nitrosylation of glucokinase, as well as vasodilation of islet vasculature. Besides, synthesis of NO is also a prerequisite for effective insulin sensitivity in targeted tissues. Thus, NO is involved in glucose uptake and disrupted NO pathways play a role in pathogenesis of insulin resistance in hypertension, obesity and type 2 diabetes mellitus. In this review, we summarize the updated paradigms on the involvement of NO in insulin secretion from islets of Langerhans and glucose uptake by various tissue system. Hence, a better understanding of nitric oxide synthase (NOS)–NO system in regulation of glucose homeostasis can hopefully facilitate the development of new treatments.
Article
Full-text available
Cardiovascular diseases (CVDs) and metabolic disorders are major components of noncommunicable diseases, causing an enormous health and economic burden worldwide. There are common risk factors and developmental mechanisms among them, indicating the far-reaching significance in exploring the corresponding therapeutic targets. MST1/2 kinases are well-established proapoptotic effectors that also bidirectionally regulate autophagic activity. Recent studies have demonstrated that MST1/2 influence the outcome of cardiovascular and metabolic diseases by regulating immune inflammation. In addition, drug development against them is in full swing. In this review, we mainly describe the roles and mechanisms of MST1/2 in apoptosis and autophagy in cardiovascular and metabolic events as well as emphasis on the existing evidence for their involvement in immune inflammation. Moreover, we summarize the latest progress of pharmacotherapy targeting MST1/2 and propose a new mode of drug combination therapy, which may be beneficial to seek more effective strategies to prevent and treat CVDs and metabolic disorders.
Article
Apoptosis repressor with caspase recruitment domain (ARC) is an established cytoplasmic anti-apoptotic factor relevant for cancer and metabolic disease. In this issue of Developmental Cell, McKimpson et al. show that ARC can assume potent pro-apoptotic effects in β cells of the endocrine pancreas via translocation to the nucleus.
Article
Full-text available
The Pyst1 and Pyst2 mRNAs encode closely related proteins, which are novel members of a family of dual‐specificity MAP kinase phosphatases typified by CL100/MKP‐1. Pyst1 is expressed constitutively in human skin fibroblasts and, in contrast to other members of this family of enzymes, its mRNA is not inducible by either stress or mitogens. Furthermore, unlike the nuclear CL100 protein, Pyst1 is localized in the cytoplasm of transfected Cos‐1 cells. Like CL100/ MKP‐1, Pyst1 dephosphorylates and inactivates MAP kinase in vitro and in vivo. In addition, Pyst1 is able to form a physical complex with endogenous MAP kinase in Cos‐1 cells. However, unlike CL100, Pyst1 displays very low activity towards the stress‐activated protein kinases (SAPKs) or RK/p38 in vitro, indicating that these kinases are not physiological substrates for Pyst1. This specificity is underlined by the inability of Pyst1 to block either the stress‐mediated activation of the JNK‐1 SAP kinase or RK/p38 in vivo, or to inhibit nuclear signalling events mediated by the SAP kinases in response to UV radiation. Our results provide the first evidence that the members of the MAP kinase family of enzymes are differentially regulated by dual‐specificity phosphatases and also indicate that the MAP kinases may be regulated by different members of this family of enzymes depending on their subcellular location.
Article
Full-text available
Stimulation of PC12 cells with nerve growth factor (NGF) increased mitogen-activated protein kinase kinase (MAPKK) activity > 20-fold after 5 min to a level that was largely sustained for at least 90 min. MAPKK activity was stimulated to a similar level by epidermal growth factor (EGF), but peaked at 2 min, declining thereafter and returning to basal levels after 60-90 min. Activation of MAPKK by either growth factor occurred prior to the activation of MAP kinase, consistent with MAPKK being the physiological activator of MAP kinase. The results demonstrate that the transient activation of MAPKK by EGF and its sustained activation by NGF underlies the transient and sustained activation of MAP kinase induced by EGF and NGF respectively. NGF or EGF induced the same two forms of MAPKK that were resolved on a Mono Q column. The Peak-1 MAPKK was activated initially and partially converted into the more acidic peak-2 MAPKK after prolonged growth-factor stimulation. The Peak-2 MAPKK was 20-fold more sensitive to inactivation by the catalytic subunit of protein phosphatase 2A. Stimulation with NGF caused a striking translocation of MAP kinase from the cytosol to the nucleus after 30 min, but not nuclear translocation of MAP kinase occurred after stimulation with EGF. The results suggest that sustained activation of the MAP kinase cascade may be required for MAP kinase to enter the nucleus, where it may initiate the gene transcription events required for neuronal differentiation of PC12 cells.
Article
Full-text available
We demonstrate that members of the erk-encoded family of mitogen-activated protein (MAP) kinases (pp44/42mapk/erk) and members of the rsk-encoded protein kinases (RSKs or pp90rsk) are present in the cytoplasm and nucleus of HeLa cells. Addition of growth factors to serum-deprived cells results in increased tyrosine and threonine phosphorylation and in the activation of cytosolic and nuclear MAP kinases. Activated MAP kinases then phosphorylate (serine/threonine) and activate RSKs. Concurrently, a fraction of the activated MAP kinases and RSKs enter the nucleus. In addition, a distinct growth-regulated RSK-kinase activity (an enzyme[s] that phosphorylates recombinant RSK in vitro and that may be another member of the erk-encoded family of MAP kinases) was found associated with a postnuclear membrane fraction. Regulation of nuclear MAP kinase and RSK activities by growth factors and phorbol ester is coordinate with immediate-early gene expression. Indeed, in vitro, MAP kinase and/or RSK phosphorylates histone H3 and the recombinant c-Fos and c-Jun polypeptides, transcription factors phosphorylated in a variety of cells in response to growth stimuli. These in vitro studies raise the possibility that the MAP kinase/RSK signal transduction pathway represents a protein-Tyr/Ser/Thr phosphorylation cascade with the spatial distribution and temporal regulation that can account for the rapid transmission of growth-regulating information from the membrane, through the cytoplasm, and to the nucleus.
Article
Full-text available
Tissue-specific expression of the human insulin gene is regulated by cis-acting DNA elements 5' to the transcription start site. Deletion of the 5' region of the human insulin gene between nucleotides -279 and -258 caused a 25-fold rise in transcriptional activity whereas further deletion to nucleotide -229 reduced transcription activity 25-fold. In vitro analysis of protein binding in the 5' regulatory region revealed: (i) the major positive regulatory region (-258 to -229) contains a protein-binding site (GC-II) with 75% sequence identity to a motif in the rat insulin I gene, shown to be a powerful transcriptional activator. GC-II motif-binding factors are not restricted to insulin-producing cell lines. (ii) An islet cell-specific factor binds between nucleotides -217 to -210 (CT-II motif). (iii) A region between nucleotides -153 and -127, containing two identical motifs, GG-I and GG-II was also revealed. GG-I-binding factors are ubiquitous, whereas binding to the GG-II motif is beta cell-specific. (iv) A ubiquitous factor binds to a motif between nucleotides -179 and -183, identical to a half-site for the cyclic nucleotide regulatory element. (v) The negative regulatory element between -279 and -258 contains overlapping binding sites for at least 3 protein factors, with differing cell-specific distributions and can independently down-regulate thymidine kinase promoter activity in a beta cell line.
Article
Full-text available
Sequence-specific binding of proteins from an insulin-secreting cell line (RINm-5F) to the human insulin-gene 5' region were examined by gel-retardation and methylation-interference analysis. Specific binding of a nuclear factor to sites between nucleotides -210 to -217 and -77 to -84 was detected. The same binding activity was shown at an upstream site (-313 to -320) with low affinity. Studies using mutated binding-site probes delineated a sequence 5'-C(T/C)CTAATG-3' for high-affinity interactions. This binding activity was also present in another insulin-producing cell line (HIT.T15), but not in extracts from cell lines that did not express the insulin gene (HeLa, HL60). Cross-species comparisons show that this sequence element is highly conserved and may thus play an important role in the cell-specific regulation of insulin-gene transcription.
Article
Guidelines for submitting commentsPolicy: Comments that contribute to the discussion of the article will be posted within approximately three business days. We do not accept anonymous comments. Please include your email address; the address will not be displayed in the posted comment. Cell Press Editors will screen the comments to ensure that they are relevant and appropriate but comments will not be edited. The ultimate decision on publication of an online comment is at the Editors' discretion. Formatting: Please include a title for the comment and your affiliation. Note that symbols (e.g. Greek letters) may not transmit properly in this form due to potential software compatibility issues. Please spell out the words in place of the symbols (e.g. replace “α” with “alpha”). Comments should be no more than 8,000 characters (including spaces ) in length. References may be included when necessary but should be kept to a minimum. Be careful if copying and pasting from a Word document. Smart quotes can cause problems in the form. If you experience difficulties, please convert to a plain text file and then copy and paste into the form.
Article
Electrophoretic mobility shift assays were performed using oligonucleotides corresponding to known protein binding sites within the human insulin gene enhancer and nuclear extracts from mouse pancreatic α and β cell lines. The results demonstrate that a previously described factor, IUF-1, binds to three sites at −82 (the CT1 box), −215 (the CT2 box), and −319 (the CT3 box) in the human insulin gene enhancer. IUF-1 was present only in β but not in α cells, while all other DNA-binding proteins were present in both cell lines. IUF-1 may therefore be an important determinant of insulin gene β cell-specific expression.
Article
An automated method for the isolation of human pancreatic islets is described. The procedure meets the following requirements: 1) minimal traumatic action on the islets, 2) continuous digestion in which the islets that are progressively liberated can be saved from further enzymatic action, 3) minimal human intervention in the digestion process, and 4) high yield and purity of the isolated islets. After purification of Ficoll gradients, an average of 2279 islets/g pancreas was obtained, with an average purity of 79% islets. The average volume and average insulin content of the final islet preparation were 348 mm3 and 93.4 U, respectively. The islets were morphologically intact with a normal degree of beta-cell granulation, responded to glucose stimulation with a fivefold increase of insulin secretion over basal levels, and produced normoglycemia after transplantation into diabetic mice. The procedure is being used in the second phase of clinical trials of islet transplantation in patients with insulin-dependent diabetes mellitus.
Article
The endocrine pancreas consists of several differentiated cell types that are distinguished by their selective expression of peptide hormones such as insulin, glucagon, and somatostatin. Although a number of homeobox-type factors have been proposed as key regulators of individual peptide genes in the pancreas, their cellular distribution and relative abundance remain uncharacterized. Also, their overlapping DNA binding specificities have further obscured the regulatory functions these factors perform during development. In this report we characterize a novel homeobox-type somatostatin transactivating factor termed STF-1, which is uniformly expressed in cells of the endocrine pancreas and small intestine. The 283-amino acid STF-1 protein binds to tissue-specific elements within the somatostatin promoter and stimulates somatostatin gene expression both in vivo and in vitro. Remarkably, STF-1 comprises the predominant tissue-specific element-binding activity in nuclear extracts from somatostatin-producing pancreatic islet cells, suggesting that this protein may have a primary role in regulating peptide hormone expression and specifying endocrine cell lineage in the developing gut.